Cyclin D1 (CCND1) is a regulatory protein essential for the G1/S phase transition of the cell cycle. Antibodies targeting Cyclin D1 are widely used in cancer research, particularly in studies of malignancies like mantle cell lymphoma and breast cancer, where Cyclin D1 overexpression is common .
Cell Signaling Technology #2922: Detects endogenous Cyclin D1 at 36 kDa. Optimal for WB and IP but requires high sensitivity protocols due to low expression in some cell types .
Proteintech 60186-1-Ig: Validated in 723+ WB studies, with confirmed reactivity in 15+ cell lines. Users report stronger signals in HeLa and A549 cells but note extended exposure times for low-abundance samples .
Cyclin D1 antibodies have been instrumental in elucidating:
Oncogenic Mechanisms: Overexpression correlates with tumor progression via dysregulation of CDK4/6 and Rb phosphorylation .
Therapeutic Targeting: Used to validate Cyclin D1 as a biomarker in preclinical models for CDK4/6 inhibitors (e.g., palbociclib) .
Structural Studies: Antibodies like 60186-1-Ig enable detection of post-translational modifications (e.g., phosphorylation at Thr286) critical for protein degradation .
Cross-Reactivity: Proteintech 60186-1-Ig shows reactivity with mouse, human, and zebrafish samples, but not with bovine or hamster tissues .
Storage: Stable at -20°C in PBS with 0.02% sodium azide. Avoid freeze-thaw cycles .
Dilution Guidelines:
Recent studies utilize Cyclin D1 antibodies in:
Single-Domain Antibody Engineering: Fusion with IgG1 Fc enhances neutralization activity (e.g., in antiviral therapies) .
Multiplex Assays: Integration with immune checkpoint inhibitors (e.g., PD-1/CTLA-4) for combinatorial cancer therapies .
CYSD1 (Cysteine Domain-containing protein 1) is a protein found in Arabidopsis thaliana that contains characteristic cysteine-rich domains. These domains share structural similarities with the CysD domains found in gel-forming mucins such as MUC2, MUC5AC, and MUC5B, which contain ten invariant cysteine residues . The CysD domains are typically found adjacent to or scattered within heavily O-glycosylated regions of proteins.
The high conservation of CysD domains across species suggests they play critical roles in protein function. While specific functions of CYSD1 in Arabidopsis remain under investigation, research into homologous CysD domains indicates they may be involved in protein dimerization and stabilization through intramolecular disulfide bonding . To study CYSD1 function, researchers typically employ genetic approaches such as gene knockouts, RNA interference, or CRISPR-Cas9 gene editing, followed by phenotypic analysis and biochemical characterization.
CYSD1 antibodies can be employed in multiple experimental applications to study protein expression, localization, and interactions. For protein detection and quantification, Western blotting represents the most common application, allowing researchers to identify CYSD1 protein in plant tissue extracts and determine relative abundance across different experimental conditions.
Immunohistochemistry and immunofluorescence techniques enable visualization of protein distribution within plant tissues and cells, providing valuable information about spatial expression patterns. Co-immunoprecipitation (Co-IP) experiments can reveal protein-protein interactions involving CYSD1 . For high-throughput screening, enzyme-linked immunosorbent assays (ELISA) offer a quantitative approach to measuring CYSD1 protein levels across multiple samples simultaneously.
When selecting application-specific protocols, researchers should optimize conditions including antibody dilution, incubation time, blocking reagents, and detection methods based on the antibody specifications and experimental goals.
Antibody validation is crucial for ensuring experimental reliability and reproducibility. For CYSD1 antibodies, multiple complementary approaches should be employed:
First, perform Western blot analysis using extracts from wild-type Arabidopsis thaliana tissues alongside extracts from CYSD1 knockout or knockdown lines. A specific antibody will show reduced or absent signal in the mutant lines compared to wild-type. Additionally, pre-incubation of the antibody with purified recombinant CYSD1 protein should block binding to the target in subsequent Western blot or immunostaining experiments.
For further validation, express tagged CYSD1 protein in a heterologous system and confirm detection using both anti-tag and anti-CYSD1 antibodies. Cross-reactivity testing should be conducted with closely related proteins, particularly those containing similar CysD domains . Lastly, mass spectrometry analysis of immunoprecipitated proteins can confirm the identity of the protein recognized by the antibody.
Immunoprecipitation (IP) of CYSD1 requires careful optimization of extraction and binding conditions. Begin by testing different extraction buffers to maintain protein solubility while preserving native interactions. A good starting point is a buffer containing 50 mM Tris-HCl (pH 7.5), 150 mM NaCl, 1% Nonidet P-40, and protease inhibitor cocktail. Consider including phosphatase inhibitors if studying phosphorylation-dependent interactions.
For antibody-based precipitation, conjugated agarose beads offer advantages over protein A/G beads with secondary antibodies, as they minimize background and improve specificity . When using agarose-conjugated CYSD1 antibodies, ensure appropriate antibody-to-lysate ratios and incubation times are established through pilot experiments. Typically, 2-5 μg antibody per 500 μg of total protein with overnight incubation at 4°C yields optimal results.
Post-translational modifications (PTMs) of CYSD1 represent an important area of investigation that requires specialized approaches. Given the cysteine-rich nature of CYSD1, redox-based modifications including disulfide bond formation and S-glutathionylation are particularly relevant. When studying these modifications, sample preparation must avoid reducing agents that would disrupt disulfide bonds.
For studying phosphorylation, use phosphatase inhibitors (sodium fluoride, sodium orthovanadate) during extraction. Immunoprecipitation with CYSD1 antibodies followed by immunoblotting with phospho-specific antibodies can reveal phosphorylation status . Alternatively, immunoprecipitated samples can be analyzed by mass spectrometry to identify specific modified residues.
If investigating glycosylation patterns of CYSD1, consider enzymatic deglycosylation assays using PNGase F or O-glycosidase followed by Western blotting to observe mobility shifts. When working with CysD domains similar to those in mucins, which are often heavily O-glycosylated , specialized glycoprotein extraction and analysis methods may be required.
It's important to note that some PTMs may affect antibody recognition. Therefore, validation experiments should confirm whether the CYSD1 antibody's epitope is accessible regardless of the protein's modification state.
Fixation and extraction methods significantly impact epitope accessibility and immunohistochemical detection of CYSD1. For plant tissues, compare paraformaldehyde (4%, 12-24 hours) versus glutaraldehyde (0.5-2.5%) fixation, as these preserve different aspects of protein structure. Paraformaldehyde generally provides better epitope preservation for antibody recognition, while glutaraldehyde offers superior ultrastructural preservation.
After fixation, antigen retrieval becomes crucial for exposing epitopes that may be masked during fixation. Heat-induced epitope retrieval (HIER) using citrate buffer (pH 6.0) or Tris-EDTA buffer (pH 9.0) at 95-100°C for 20-30 minutes often improves staining intensity. Enzymatic retrieval using proteinase K may be beneficial for certain fixation conditions but carries the risk of excessive tissue digestion.
Comprehensive analysis of CYSD1 expression requires a multi-method approach. Begin with quantitative RT-PCR to measure transcript levels across different tissues (roots, stems, leaves, flowers) and developmental stages. Design gene-specific primers spanning exon-exon junctions to avoid genomic DNA amplification, and normalize expression to multiple reference genes appropriate for the experimental conditions.
For protein-level analysis, perform Western blotting on tissue-specific extracts using CYSD1 antibodies . Quantify band intensities relative to loading controls such as actin or GAPDH. Tissue-specific differences in protein extraction efficiency should be accounted for when interpreting results.
Immunohistochemistry or immunofluorescence provides spatial information about CYSD1 distribution. For Arabidopsis, consider both whole-mount immunostaining for seedlings and section-based approaches for mature tissues. To complement antibody-based methods, generate transgenic plants expressing CYSD1-reporter fusions (GFP or mCherry) under native promoter control.
For highest resolution analysis, laser capture microdissection can isolate specific cell types for CYSD1 expression analysis. This approach allows correlation of expression patterns with specific cellular functions across developmental stages.
Robust co-immunoprecipitation (Co-IP) experiments require comprehensive controls to distinguish genuine interactions from artifacts. Primary controls must include:
Input control: Analysis of pre-immunoprecipitation lysate to confirm target protein expression and establish a reference for enrichment calculation.
Negative antibody control: Parallel immunoprecipitation using non-immune IgG of the same isotype and concentration as the CYSD1 antibody to identify non-specific binding.
Genetic controls: Compare Co-IP results from wild-type plants with those from CYSD1 knockout or knockdown lines to confirm specificity.
Reciprocal Co-IP: When a potential interaction partner is identified, confirm the interaction by immunoprecipitating with antibodies against the partner and detecting CYSD1 in the precipitate.
Additional validation steps should include testing interaction stability under different salt concentrations (150-500 mM NaCl) to assess binding strength and specificity. For suspected indirect interactions, include RNase and DNase treatments during immunoprecipitation to exclude nucleic acid-mediated co-precipitation. Finally, confirm biological relevance of identified interactions through functional assays such as genetic epistasis analysis or co-localization studies.
Discrepancies between antibody-based detection and genomic/transcriptomic data require systematic troubleshooting. First, examine antibody specificity through Western blotting with recombinant CYSD1 protein and knockout controls to rule out non-specific recognition. Consider epitope accessibility issues that may affect antibody binding under certain experimental conditions.
Post-transcriptional regulation may explain differences between mRNA and protein levels. Investigate potential microRNA-mediated regulation or alternative splicing events that might affect CYSD1 expression. Protein stability and turnover rate also influence steady-state protein levels independent of transcription. Pulse-chase experiments with protein synthesis inhibitors can reveal differences in CYSD1 protein half-life across tissues or conditions.
Subcellular localization differences may lead to apparent contradictions if fractionation methods used in Western blotting exclude relevant cellular compartments. Perform comprehensive subcellular fractionation to track CYSD1 distribution across cellular compartments.
For quantitative discrepancies, consider technical limitations in detection sensitivity. RNA-seq or qRT-PCR can detect low-abundance transcripts that may not produce enough protein for antibody detection. Conversely, stable proteins may accumulate to detectable levels even with low transcript abundance.
CysD domains exhibit significant conservation across species, suggesting important functional roles. These domains have been identified in gel-forming mucins from mammals, frog, fish, fruit fly, sea urchin, sea squirt, and lancelet . The high intra- and inter-species homologies of CysD domains reflect evolutionary conservation of essential structural elements.
When considering cross-species application of CYSD1 antibodies, alignment of the antibody epitope sequence across target species is essential. If the epitope region shows high conservation (>80% sequence identity), cross-reactivity is possible but must be experimentally verified. Perform Western blot analysis using protein extracts from target species alongside positive controls from Arabidopsis thaliana.
Interestingly, CysD domains show homology with domains in human cartilage intermediate layer protein and in Oikosin 1 from the larvacean tunicate Oikopleura dioica . This broad conservation suggests functional importance across diverse organisms and provides opportunities for comparative studies of CysD domain-containing proteins.
For comprehensive cross-species studies, complementary approaches such as epitope tagging of CYSD1 orthologs may be necessary when antibody cross-reactivity is insufficient.
Plant CYSD1 shares structural similarities with mammalian mucin CysD domains, particularly the characteristic pattern of conserved cysteine residues. In mammalian mucins like MUC2, MUC5AC, and MUC5B, CysD domains consist of approximately 110 residues with ten invariant cysteine residues . These domains are typically found adjacent to or within heavily O-glycosylated regions of the proteins.
A key functional property of mammalian CysD domains is their ability to form pH-independent non-covalent dimers stabilized by intramolecular disulfide bonds . This dimerization property contributes to the gel-forming characteristics of mucins. While plant CYSD1 contains similar cysteine-rich domains, its specific oligomerization properties may differ from mammalian counterparts.
Another distinction involves glycosylation patterns. Mammalian mucin CysD domains are associated with heavily O-glycosylated PTS (proline, threonine, serine-rich) domains . Plant CYSD1 may have different glycosylation patterns reflecting the distinct glycosylation machinery in plants compared to mammals.
Despite these differences, the evolutionary conservation of these domains suggests fundamental structural or functional importance. Comparative biochemical studies examining the disulfide bonding patterns and interaction properties of plant CYSD1 versus mammalian CysD domains would provide valuable insights into their respective biological roles.
Machine learning (ML) offers transformative potential for CYSD1 antibody engineering and application. Recent advances in ML-guided antibody engineering employ a "lab-in-a-loop" approach with iterative cycles of computational design and experimental validation . This approach can optimize complementarity-determining regions (CDRs) to enhance binding affinity and select appropriate scaffolds for improved developability.
For CYSD1 antibody development, machine learning models like AbRFC can predict non-deleterious mutations in antibody CDRs, allowing targeted screening of a manageable number of candidate mutations (approximately 100) to identify those with enhanced properties . This approach has demonstrated significant affinity improvements with limited experimental screening.
The second iteration in ML-guided antibody engineering involves CDR-FR shuffling, combining optimized CDR loops with diverse human VH/VL frameworks to generate clinically developable candidates with enhanced pharmacokinetic properties . This approach is particularly valuable for developing antibodies suitable for multiple applications.
Implementation of ML approaches requires careful consideration of training datasets. Despite limitations in publicly available training data, classical ML remains effective for predicting the impact of small molecular changes on function and properties . As experimental datasets grow through iterative antibody engineering campaigns, the predictive performance of ML models improves, creating a positive feedback loop for future antibody development efforts.
Single-domain antibodies (sdAbs) represent an emerging technology with significant potential for CYSD1 research. These compact antibody fragments, derived from heavy-chain-only antibodies found in camelids or engineered from conventional antibodies, offer advantages including small size, high stability, and access to epitopes that conventional antibodies cannot reach.
Recent advances have demonstrated the development of humanized single domain antibodies with high binding affinity, exemplified by sdAbs against SARS-CoV-2 spike protein receptor-binding domain with equilibrium dissociation constants (KD) in the nanomolar range (0.99-35.5 nM) . Similar approaches could be applied to generate high-affinity sdAbs against CYSD1.
The monomeric nature of sdAbs provides excellent tissue penetration, while their single-domain architecture simplifies recombinant production in various expression systems. For enhanced functionality, sdAbs can be fused to Fc domains, improving their neutralization activity by up to ten times as demonstrated in viral neutralization studies .
For CYSD1 research, sdAbs could enable super-resolution microscopy studies through site-specific labeling, facilitate intracellular tracking when expressed as intrabodies, and allow precise manipulation of protein function through targeted binding to specific functional domains. Additionally, sdAbs can be formatted into multispecific molecules to study CYSD1 interactions with other proteins simultaneously.
CRISPR-Cas genome editing technologies are revolutionizing CYSD1 research by enabling precise genetic modifications that enhance antibody application and validation. Knockout lines created through CRISPR provide the gold standard negative controls for antibody specificity validation. These lines can be generated with minimal off-target effects and without the positional effects associated with T-DNA insertion mutants.
Epitope tagging through CRISPR-mediated homology-directed repair allows endogenous tagging of CYSD1 with small epitope tags or fluorescent proteins at either terminus or at internal sites. This approach preserves native expression patterns and regulatory mechanisms while enabling detection with well-characterized anti-tag antibodies when specific CYSD1 antibodies are unavailable or problematic.
CRISPR interference (CRISPRi) and CRISPR activation (CRISPRa) systems provide tunable control over CYSD1 expression without permanent genetic modification. These systems are valuable for distinguishing between primary and secondary effects of CYSD1 perturbation through temporal control of expression changes.
Base editing and prime editing technologies allow introduction of specific point mutations in CYSD1 to study structure-function relationships, particularly the role of individual cysteine residues in disulfide bond formation and protein function. These precise modifications facilitate detailed antibody epitope mapping studies and help resolve questions about antibody recognition of specific protein variants or modified forms.